Tian-Tian Sun
Shanghai Jiao Tong University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Tian-Tian Sun.
Cancer Letters | 2013
Shuai Wu; Xin-Yuan Lao; Tian-Tian Sun; Lin-Lin Ren; Xuan Kong; Ji-Lin Wang; Ying-Chao Wang; Wan Du; Ya-Nan Yu; Yu-Rong Weng; Jie Hong; Jing-Yuan Fang
Zinc finger protein X-linked (ZFX) is a zinc finger transcription factor encoded on the X chromosome. Here, we found that ZFX expression was significantly upregulated in gastric cancer (GC) cell lines and tissues. Knockdown of ZFX induced significant apoptosis and cell cycle arrest in SGC7901 and MGC803 cells. Moreover, we demonstrated for the first time that knockdown of ZFX inhibited gastric cancer cell growth in vitro and in vivo via downregulating the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK-MAPK) pathway. Therefore, ZFX play a prominent role in GC tumorigenicity and may have potential applications in the diagnosis or treatment of GC.
Cell Death and Disease | 2014
Ji-Lin Wang; Chen Zf; Hui-Min Chen; Wang My; Xuan Kong; Yuyao Wang; Tian-Tian Sun; Jie Hong; Zou W; Jianrong Xu; Jing-Yuan Fang
Aberrant regulation of the Wnt/β-catenin pathway plays important roles in colorectal carcinogenesis, with over 90% of cases of sporadic colon cancer featuring β-catenin accumulation. While ubiquitination-mediated degradation is widely accepted as a major route for β-catenin protein turnover, little is known about the regulation of β-catenin in transcriptional level. Here we show that Elf3, a member of the E-twenty-six family of transcription factors, drives β-catenin transactivation and associates with poor survival of colorectal cancer (CRC) patients. We first found recurrent amplification and upregulation of Elf3 in CRC tissues, and further Gene Set Enrichment Analysis identified significant association between Elf3 expression and activity of WNT/β-catenin pathway. Chromatin immunoprecipitation and electrophoretic mobility shift assay consistently revealed that Elf3 binds to and transactivates β-catenin promoter. Ectopic expression of Elf3 induces accumulation of β-catenin in both nucleus and cytoplasm, causing subsequent upregulation of several effector genes including c-Myc, VEGF, CCND1, MMP-7 and c-Jun. Suppressing Elf3 in CRC cells attenuates β-catenin signaling and decreases cell proliferation, migration and survival. Targeting Elf3 in xenograft tumors suppressed tumor progression in vivo. Taken together, our data identify Elf3 as a pivotal driver for β-catenin signaling in CRC, and highlight potential prognostic and therapeutic significance of Elf3 in CRC.
Oncotarget | 2015
Ya-Nan Yu; Ta-Chung Yu; Hui-Jun Zhao; Tian-Tian Sun; Hui-Min Chen; Haoyan Chen; Hui-Fang An; Yu-Rong Weng; Jun Yu; Min Li; Wen-Xin Qin; Xiong Ma; Nan Shen; Jie Hong; Jing-Yuan Fang
Background Accumulating evidence links colorectal cancer (CRC) with the intestinal microbiota. However, the disturbance of intestinal microbiota and the role of Fusobacterium nucleatum during the colorectal adenoma-carcinoma sequence have not yet been evaluated. Methods 454 FLX pyrosequencing was used to evaluate the disturbance of intestinal microbiota during the adenoma-carcinoma sequence pathway of CRC. Intestinal microbiota and mucosa tumor-immune cytokines were detected in mice after introducing 1,2-dimethylhydrazine (DMH), F. nucleatum or Berberine (BBR), using pyrosequencing and Bio-Plex Pro™ cytokine assays, respectively. Protein expressions were detected by western blotting. Results The levels of opportunistic pathogens, such as Fusobacterium, Streptococcus and Enterococcus spp. gradually increased during the colorectal adenoma-carcinoma sequence in human fecal and mucosal samples. F. nucleatum treatment significantly altered lumen microbial structures, with increased Tenericutes and Verrucomicrobia (opportunistic pathogens) (P < 0.05 = in wild-type C57BL/6 and mice with DMH treatment). BBR intervention reversed the F. nucleatum-mediated increase in opportunistic pathogens, and the secretion of IL-21/22/31, CD40L and the expression of p-STAT3, p-STAT5 and p-ERK1/2 in mice, compared with mice fed with F. nucleatum alone. Conclusions F. nucleatum colonization in the intestine may prompt colorectal tumorigenesis. BBR could rescue F. nucleatum-induced colorectal tumorigenesis by modulating the tumor microenvironment and blocking the activation of tumorigenesis-related pathways.
International Journal of Cancer | 2014
Hua Xiong; Wan Du; Tian-Tian Sun; Yan-Wei Lin; Ji-Lin Wang; Jie Hong; Jing-Yuan Fang
Persistent infection with Helicobacter pylori (H. pylori) contributes to gastric diseases including chronic gastritis and gastric cancer. However, the pathogenesis of this carcinogenic bacterium has not been completely elucidated. Here, we report that H. pylori rapidly triggers STAT3 signaling and induces STAT3‐dependent COX‐2 expression both in vitro and in vivo. STAT3 upregulats COX‐2 by binding to and increasing the activity of COX‐2 promoter. COX‐2 in turn regulates IL‐6/STAT3 signaling under basal conditions and during H. pylori infection. These findings suggest that a positive feedback loop between STAT3 and COX‐2 exists in the basal condition and H. pylori infectious condition. Immunohistochemical staining revealed that H. pylori‐positive gastritis tissues exhibited markedly higher levels of pSTAT3Tyr705 than H. pylori‐negative ones. High pSTAT3Tyr705 levels are correlated with intestinal metaplasia and dysplasia, suggesting pSTAT3Tyr705 may be useful in the early detection of gastric tumorigenesis. Additionally, a strong positive correlation between STAT3/pSTAT3Tyr705 levels and COX‐2 expression was identified in gastritis and gastric cancer tissues. Together, these findings provide new evidence for a positive feedback loop between STAT3 signaling and COX‐2 in H. pylori pathogenesis and may lead to new approaches for early detection and effective therapy of gastric cancer.
Apoptosis | 2014
Ying-Chao Wang; Ji-Lin Wang; Xuan Kong; Tian-Tian Sun; Haoyan Chen; Jie Hong; Jing-Yuan Fang
The development of gastric cancer (GC) is a complex multistep process, including numerous genetic and epigenetic changes. CD24 is associated with enhanced invasiveness of GC and a poor prognosis. However, the mechanism by which CD24 induces GC progression remains poorly characterized. Here, we found that the expression of CD24 gradually increased in samples of normal gastric mucosa, non-atrophic chronic gastritis, chronic atrophic gastritis (CAG), CAG with intestinal metaplasia, dysplasia and GC. Moreover, the knockdown of CD24 induced significant levels of apoptosis in GC cells via the mitochondrial apoptotic pathway. CD24 may also promote cellular invasion and regulate the expression of E-cadherin, fibronectin and vitamin D receptor in GC cells. The activation of signal transducer and activator of transcription 3 (STAT3) may mediate CD24-induced GC survival and invasion in vitro. Furthermore, CD24-induced GC progression and STAT3 activation could also be detected in vivo and in clinical GC tissues samples. Taken together, our results indicate that CD24 mediates gastric carcinogenesis and may promote GC progression by suppressing apoptosis and promoting invasion, with the activation of STAT3 playing a critical role.
Clinical Cancer Research | 2014
Tian-Tian Sun; Wan Du; Hua Xiong; Ya-Nan Yu; Yu-Rong Weng; Lin-Lin Ren; Hui-Jun Zhao; Ying-Chao Wang; Ying-Xuan Chen; Jie Xu; Yongbing Xiang; Wen-Xin Qin; Weibiao Cao; Weiping Zou; Haoyan Chen; Jie Hong; Jing-Yuan Fang
Purpose: The role and clinical implication of the transmembrane protein with EGF and two follistatin motifs 2 (TMEFF2) in gastric cancer is poorly understood. Experimental Design: Gene expression profile analyses were performed and Gene Set Enrichment Analysis (GSEA) was used to explore its gene signatures. AGS and MKN45 cells were transfected with TMEFF2 or control plasmids and analyzed for gene expression patterns, proliferation, and apoptosis. TMEFF2 expression was knocked down with shRNAs, and the effects on genome stability were assessed. Interactions between TMEFF2 and SHP-1 were determined by mass spectrometry and immunoprecipitation assays. Results: Integrated analysis revealed that TMEFF2 expression was significantly decreased in gastric cancer cases and its expression was negatively correlated with the poor pathologic stage, large tumor size, and poor prognosis. GSEA in The Cancer Genome Atlas (TCGA) and Jilin datasets revealed that cell proliferation, apoptosis, and DNA damage–related genes were enriched in TMEFF2 lower expression patients. Gain of TMEFF2 function decreased cell proliferation by increasing of apoptosis and blocking of cell cycle in gastric cancer cells. The protein tyrosine phosphatase SHP-1 was identified as a binding partner of TMEEF2 and mediator of TMEFF2 function. TMEFF2 expression positively correlated with SHP-1, and a favorable prognosis was more likely in patients with gastric cancer with higher levels of both TMEFF2 and SHP-1. Conclusion: TMEFF2 acts as a tumor suppressor in gastric cancer through direct interaction with SHP-1 and can be a potential biomarker of carcinogenesis. Clin Cancer Res; 20(17); 4689–704. ©2014 AACR.
International Journal of Cancer | 2015
Tian-Tian Sun; Jia-Yin Tang; Wan Du; Hui-Jun Zhao; Gang Zhao; Shengli Yang; Haoyan Chen; Jie Hong; Jing-Yuan Fang
The transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is a single‐pass transmembrane protein, and it is downregulated in human gastric cancer and levels correlate with tumor progression and time of survival. However, the mechanism of its dysregulation in gastric cancer is little known. Here we investigate its regulatory mechanism and the bidirectional regulation between TMEFF2 and STAT3 in gastric carcinogenesis. TMEFF2 expression was decreased after Helicobacter pylori (H. pylori) infection in vivo and in vitro. STAT3 directly binds to the promoter of TMEFF2 and regulates H. pylori‐induced TMEFF2 downregulation in normal gastric GES‐1 cells and gastric cancer AGS cells. Conversely, TMEFF2 may suppress the phosphorylation of STAT3 and TMEFF2‐induced downregulation of STAT3 phosphorylation may depend on SHP‐1. A highly inverse correlation between the expression of TMEFF2 and pSTAT3 was also revealed in gastric tissues. We now show the deregulation mechanism of TMEFF2 in gastric carcinogenesis and identify TMEFF2 as a new target gene of STAT3. The phosphorylation of STAT3 may be negatively regulated by TMEFF2, and the bidirectional regulation between TMEFF2 and STAT3 may contribute to H. pylori‐associated gastric carcinogenesis.© 2014 UICC
Clinical Cancer Research | 2017
Qian Liang; Dan Ma; Xiaoqiang Zhu; Zhen-Hua Wang; Tian-Tian Sun; Chaoqin Shen; Tingting Yan; Xianglong Tian; TaChuang Yu; Fangfang Guo; Jia-Yin Tang; Yan-Wei Lin; Hui-Min Chen; Chao Zhou; Zhizheng Ge; Ming Zhong; Jinxian Chen; Qiang Liu; Zheng Wang; Jing-Yuan Fang; Haoyan Chen; Jie Hong
Objective: The E3 ubiquitin ligase RNF6 (RING-finger protein 6) plays a crucial role in carcinogenesis. However, the copy number and expression of RNF6 were rarely reported in colorectal cancer. We aimed to explore the mechanical, biological, and clinical role of RNF6 in colorectal cancer initiation and progression. Design: The copy number and expression of RNF6 were analyzed from Tumorscape and The Cancer Genome Atlas (TCGA) datasets. Gene expressions were examined by real-time PCR, Western blot, and immunohistochemical staining. Gene expression profiling studies were performed to identify pivotal genes regulated by RNF6. Biological function of RNF6 on tumor growth and metastasis was detected in vivo and in vitro. Role of RNF6 in modulating SHP-1 expression was examined by coimmunoprecipitation and confocal microscopy, respectively. Results: The copy number of RNF6 was significantly amplified in colorectal cancer, and the amplification was associated with RNF6 expression level. Amplification and overexpression of RNF6 positively correlated with patients with colorectal cancer with poor prognosis. The gene set enrichment analysis (GSEA) revealed cell proliferation, and invasion-related genes were enriched in RNF6 high-expressed colorectal cancer cells as well as in patients from TCGA dataset. Downregulation of RNF6 impaired the colorectal cancer cell proliferation and invasion in vitro and in vivo. RNF6 may activate the JAK/STAT3 pathway and increase pSTAT3 levels by inducing the ubiquitination and degradation of SHP-1. Conclusions: Genomic amplification drives RNF6 overexpression in colorectal cancer. RNF6 may be a novel biomarker in colorectal carcinogenesis, and RNF6 may increase pSTAT3 level via promoting SHP-1 ubiquitylation and degradation. Targeting the RNF6/SHP-1/STAT3 axis provides a potential therapeutic option for RNF6-amplified tumors. Clin Cancer Res; 24(6); 1473–85. ©2017 AACR.
Molecular Oncology | 2018
Xiaoqiang Zhu; Xianglong Tian; Tian-Tian Sun; Chenyang Yu; Yingying Cao; Tingting Yan; Chaoqin Shen; Yan-Wei Lin; Jing-Yuan Fang; Jie Hong; Haoyan Chen
Although several prognostic signatures have been developed for gastric cancer (GC), the utility of these tools is limited in clinical practice due to lack of validation with large and multiple independent cohorts, or lack of a statistical test to determine the robustness of the predictive models. Here, a prognostic signature was constructed using a least absolute shrinkage and selection operator (LASSO) Cox regression model and a training dataset with 300 GC patients. The signature was verified in three independent datasets with a total of 658 tumors across multiplatforms. A nomogram based on the signature was built to predict disease‐free survival (DFS). Based on the LASSO model, we created a GeneExpressScore signature (GESGC) classifier comprised of eight mRNA. With this classifier patients could be divided into two subgroups with distinctive prognoses [hazard ratio (HR) = 4.00, 95% confidence interval (CI) = 2.41–6.66, P < 0.0001]. The prognostic value was consistently validated in three independent datasets. Interestingly, the high‐GESGC group was associated with invasion, microsatellite stable/epithelial–mesenchymal transition (MSS/EMT), and genomically stable (GS) subtypes. The predictive accuracy of GESGC also outperformed five previously published signatures. Finally, a well‐performed nomogram integrating the GESGC and four clinicopathological factors was generated to predict 3‐ and 5‐year DFS. In summary, we describe an eight‐mRNA‐based signature, GESGC, as a predictive model for disease progression in GC. The robustness of this signature was validated across patient series, populations, and multiplatform datasets.
The Journal of Pathology | 2013
Yan-Wei Lin; Lin-Lin Ren; Hua Xiong; Wan Du; Ya-Nan Yu; Tian-Tian Sun; Yu-Rong Weng; Zhen-Hua Wang; Ji-Lin Wang; Ying-Chao Wang; Yun Cui; Dan-Feng Sun; Ze-Guang Han; Nan Shen; Weiping Zou; Jie Xu; Haoyan Chen; Weibiao Cao; Jie Hong; Jing-Yuan Fang
The above article from The Journal of Pathology, published online on 7 June 2013 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the authors, the journal Editor‐in‐Chief, Prof. C. Simon Herrington, and John Wiley & Sons Limited. Some sequences in Figure 6C were mistakenly identified, with consequent errors in the description of this figure and in the Materials and Methods section. Additionally, in Figures 5C, 5F and 7 some images were duplicated and erroneously presented as unique. The authors apologise to readers of the journal.